24 Lipoic Acid

α-Lipoic acid (LA), also known as thioctic acid, is a naturally occurring compound that is synthesized in small amounts by plants and animals, including humans.1,2 Endogenously synthesized LA is covalently bound to specific proteins, which function as cofactors for several important mitochondrial enzyme complexes (see the Biological Activities section below). In addition to the physiological functions of protein-bound LA, there is increasing scientific and medical interest in potential therapeutic uses of pharmacological doses of free LA.3 LA contains two thiol (sulfur) groups, which may be oxidized or reduced (Fig. 24.1). The reduced form is known as dihydrolipoic acid (DHLA), while the oxidized form is known as LA.4 LA also contains an asymmetric carbon, meaning there are two possible optical isomers that are mirror images of each other (R- LA and S-LA). Only the R-isomer is endogenously synthesized and bound to protein; R-LA occurs naturally in foods (see the Food Sources section below). Free LA supplements may contain either R-LA or a 50/50 (racemic) mixture of R-LA and S-LA (see the Supplements section below).

Bioavailability and Metabolism

Endogenous Biosynthesis

LA is synthesized de novo from an eight-carbon fatty acid (octanoic acid) in mitochondria, where protein-bound LA functions as an enzyme cofactor. Evidence suggests that LA can be synthesized “on site” from octanoic acid that is already covalently bound to LA-dependent enzymes.5,6 The final step in LA synthesis is the insertion of two sulfur atoms into octanoic acid. This reaction is catalyzed by lipoyl synthase, an enzyme that contains iron–sulfur clusters, which are thought to act as sulfur donors to LA.7,8

Dietary and Supplemental α-Lipoic Acid

Exogenous LA from the diet can be activated with adenosine triphosphate (ATP) or guanosine triphosphate (GTP) by lipoate-activating enzyme, and transferred to LA-dependent enzymes by lipoyltransferase.9,10 Consumption of LA from food has not yet been found to result in detectable increases of free LA in human plasma or cells.3,11 In contrast, high oral doses of free LA (>50 mg) result in significant but transient increases in free LA in plasma and cells. Pharmacokinetic studies in humans have found that approximately 30%–40% of an oral dose of racemic LA is absorbed.11,12 Oral LA supplements are better absorbed on an empty stomach than with food: taking racemic LA with food decreased peak plasma LA concentrations by approximately 30% and total plasma LA concentrations by approximately 20% compared with fasting.13 Additionally, the sodium salt of R-LA may be better absorbed than free LA, presumably because of its higher aqueous solubility.14

image

Fig. 24.1 Chemical structure of lipoic acid. *Lipoic acid has a chiral center, which means it can be found in two mirror image forms (S- and R-alpha-lipoic acid) that cannot be superimposed on each other.

There may also be differences in the bioavailability of the two isomers of LA. After oral dosing with racemic LA (a 50/50 mixture of R-LA and S- LA), peak plasma concentrations of R-LA were found to be 40%–50% higher than those of S-LA, suggesting that R-LA is better absorbed than S- LA.12,14,15 Following oral administration, both isomers are rapidly metabolized and excreted. Plasma LA concentrations generally peak in 1 hour or less and decline rapidly.11,12,15,16 In cells, LA is quickly reduced to DHLA, and in vitro studies indicate that DHLA is rapidly exported from cells.3

Biological Activities

Protein-Bound α-Lipoic Acid

Enzyme Cofactor

R-LA is an essential cofactor for several mitochondrial enzyme complexes that catalyze critical reactions related to energy production and the catabolism of α-keto acids and amino acids.17 In each case, R-LA is covalently bound to a specific lysine residue in one of the proteins of the enzyme complex. The pyruvate dehydrogenase complex catalyzes the conversion of pyruvate to acetyl-coenzyme A (CoA), an important substrate for energy production via the tricarboxylic acid (TCA) cycle. The α-ketoglutarate dehydrogenase complex catalyzes the conversion of α-ketoglutarate to succinyl CoA, another important intermediate of the TCA cycle. The activity of the branchedchain α-ketoacid dehydrogenase complex results in the catabolism of the branchedchain amino acids: leucine, isoleucine, and valine.18 The glycine cleavage system is a multi-enzyme complex that catalyzes the oxidation of glycine to form 5,10-methylene tetrahydrofolate, an important cofactor in the synthesis of nucleic acids.19

Free α-Lipoic Acid

When considering the biological activities of supplemental free LA, it is important to keep in mind the limited and transient nature of the increases in plasma and tissue LA (see the Bioavailability and Metabolism section above).3

Antioxidant Activities

Scavenging reactive oxygen and nitrogen species. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are highly reactive compounds with the potential to damage DNA, proteins, and lipids (fats) in cell membranes. Both LA and DHLA can directly scavenge (neutralize) physiologically relevant ROS and RNS in the test tube.3 However, it is not clear whether LA acts directly to scavenge ROS and RNS in vivo. The highest tissue concentrations of free LA likely to be achieved through oral supplementation are at least 10 times lower than those of other intracellular antioxidants, such as vitamin C and glutathione. Moreover, free LA is rapidly eliminated from cells, so any increases in direct radical-scavenging activity are unlikely to be sustained.

Regeneration of other antioxidants. When an antioxidant scavenges a free radical, it becomes oxidized itself and is not able to scavenge additional ROS or RNS until it has been reduced. DHLA is a potent reducing agent with the capacity to reduce the oxidized forms of several important antioxidants, including vitamin C and glutathione.20 DHLA may also reduce the oxidized form of α-tocopherol (the α-tocopheroxyl radical), directly or indirectly, by reducing the oxidized form of vitamin C (dehydroascorbate), which is able to reduce the α-tocopheroxyl radical.21 Co-enzyme Q10, an important component of the mitochondrial electron transport chain, also has antioxidant activity. DHLA can reduce oxidized forms of coenzyme Q10,22 which may reduce the α-tocopheroxyl radical.23 Although DHLA has been found to regenerate oxidized antioxidants in the test tube, it is not known whether DHLA effectively regenerates other antioxidants under physiological conditions.3

Metal chelation. Redox-active metal ions, such as free iron and copper, can induce oxidative damage by catalyzing reactions that generate highly reactive free radicals.24 Compounds that chelate (bind) free metal ions in a way that prevents them from generating free radicals offer promise in the treatment of neurodegenerative diseases and other chronic diseases in which metal-induced oxidative damage may play a pathogenic role.25 Both LA and DHLA have been found to inhibit copper- and iron-mediated oxidative damage in the test tube26,27 and excess iron and copper accumulation in animal models.28,29

Induction of glutathione synthesis. Glutathione is an important intracellular antioxidant that also plays a role in the detoxification and elimination of potential carcinogens and toxins. Studies in rodents have found that glutathione synthesis and tissue glutathione levels are significantly lower in aged animals compared with younger animals, leading to decreased ability of aged animals to respond to oxidative stress or toxin exposure.30 LA has been found to increase glutathione levels in cultured cells and in the tissues of aged animals fed LA.31,32 Research suggests that LA may increase glutathione synthesis in aged rats by increasing the expression of γ-glutamylcysteine ligase (GCL), the rate-limiting enzyme in glutathione synthesis33 and by increasing cellular uptake of cysteine, an amino acid required for glutathione synthesis.34

Modulation of Signal Transduction

Insulin signaling. The binding of insulin to the insulin receptor (IR) triggers the autophosphorylation of several tyrosine residues on the IR. Activation of the IR in this manner stimulates a cascade of protein phosphorylations, resulting in the translocation of glucose transporters (GLUT4) to the cell membrane and thus increased cellular glucose uptake.3,35 LA has been found to activate the insulin signaling cascade in cultured cells,3,35,36 increase GLUT4 translocation to cell membranes, and increase glucose uptake in cultured adipose (fat) and muscle cells.37,38 A computer modeling study showed that LA binds to the tyrosine kinase domain of the IR and may stabilize the active form of the enzyme.36

PKB/Akt-dependent signaling. In addition to insulin signaling, phosphorylation and dephosphorylation of other cell-signaling molecules affect a variety of cellular processes, including metabolism, stress responses, proliferation, and survival.3 One such molecule is protein kinase B, also known as Akt (PKB/Akt). LA has been found to activate PKB/Akt-dependent signaling in vitro36,3941 and in vivo,41 inhibit apoptosis in cultured hepatocytes,36 and increase survival of cultured neurons.39 LA has also been shown to improve nitric oxide–dependent vasodilation in aged rats by increasing PKB/Akt-dependent phosphorylation of endothelial nitric oxide synthase (eNOS), which increases eNOS-catalyzed production of nitric oxide.42

Redox-sensitive transcription factors. Transcription factors are proteins that bind to specific sequences of DNA and promote or repress the transcription of selected genes. Some transcription factors are sequestered outside the nucleus until some sort of signal induces their translocation to the nucleus. Oxidative stress or changes in the balance between oxidation and reduction (redox status) in a cell can trigger the translocation of redox-sensitive transcription factors to the nucleus. One such redox-sensitive transcription factor, known as nuclear factor kappa B (NF-κB), regulates several genes related to inflammation and cell-cycle control, that are involved in the pathology of diabetes, atherosclerosis, and cancer.19 Physiologically relevant concentrations of LA added to cultured cells have been found to inhibit NF-κB nuclear translocation.43 Another redox-sensitive transcription factor known as Nrf2 enhances the transcription of genes that contain specific DNA sequences known as antioxidant response elements (AREs). LA has been found to enhance the nuclear translocation of Nrf2 and the transcription of genes containing AREs in vivo, including genes for GCL, the rate-limiting enzyme in glutathione synthesis.33

Deficiency

LA deficiency has not been described, suggesting that humans are able to synthesize enough to meet their needs for enzyme cofactors.44

Disease Treatment

Diabetes Mellitus

Chronically elevated blood glucose levels are the hallmark of diabetes mellitus (DM). In type 1 DM, insulin production is insufficient due to autoimmune destruction of the insulin-producing β-cells of the pancreas. Type 1 DM is also known as insulin-dependent DM because exogenous insulin is required to maintain normal blood glucose levels. In contrast, impaired cellular glucose uptake in response to insulin (insulin resistance) plays a key role in the development of type 2 DM.45 Although individuals with type 2 DM may eventually require insulin, type 2 DM is known as noninsulin-dependent DM because interventions that enhance insulin sensitivity may be used to maintain normal blood glucose levels.

Glucose Utilization

There is limited evidence that high doses of LA can improve glucose utilization in individuals with type 2 DM. A small clinical trial in 13 patients with type 2 DM found that a single intravenous infusion of 1000 mg of racemic LA improved insulin-stimulated glucose disposal (insulin sensitivity) by 50% compared with a placebo infusion.46 In an uncontrolled pilot study of 20 patients with type 2 DM, intravenous infusion of 500 mg/day of racemic LA for 10 days also improved insulin sensitivity when measured 24 hours after the last infusion.47 A placebo-controlled study of 72 patients with type 2 DM found that oral administration of racemic LA at doses of 600 mg/day, 1200 mg/day, or 1800 mg/day improved insulin sensitivity by 25% after 4 weeks of treatment.48 There were no significant differences among the three doses of LA, suggesting that 600 mg/day may be the maximum effective dose.45 Data from animal studies suggest that the R-isomer of LA may be more effective in improving insulin sensitivity than the S-isomer,38,49 but this possibility has not been tested in any published human trials.

The effect of LA supplementation on long-term blood glucose (glycemic) control has not been well studied. In an uncontrolled pilot study of a controlled-release form of oral racemic LA, 15 patients with type 2 DM took 900 mg/day for 6 weeks and 1200 mg/day for another 6 weeks, in addition to their current medications.15 At the end of 12 weeks, plasma fructosamine concentrations decreased by approximately 10% from baseline, but glycosylated hemoglobin (HbA1c) levels did not change. Plasma fructosamine levels reflect blood glucose control over the past 2–3 weeks, while HbA1c values reflect blood glucose control over the past 2–4 months. At present, it is not clear whether oral or intravenous LA therapy improves long-term glycemic control in individuals with type 2 DM.

Vascular Disease

The inner lining of blood vessels, known as the endothelium, plays an important role in vascular disease. Endothelial function is often impaired in patients with diabetes, who are at high risk for vascular disease.50 Intraarterial infusion of racemic LA improved endothelium-dependent vasodilation in 39 patients with diabetes but not in 11 healthy controls.51 In addition, a randomized, double-blind, placebo-controlled study in 30 patients with type 2 diabetes found that intravenous infusion of 600 mg of LA improved the response to the endothelium-dependent vasodilator acetylcholine, but not to the endothelium-independent vasodilator, glycerol trinitrate.52 Endothelial function can be assessed noninvasively by using ultrasound to measure flow-mediated vasodilation, which is endothelium dependent.53 Using ultrasound, intravenous LA has also been shown to improve endothelial function in patients with impaired fasting glucose54 or impaired glucose tolerance,55 which are prediabetic conditions.

A few studies have investigated whether oral administration of LA might improve vascular function in patients with diabetes or metabolic syndrome. A randomized controlled trial assessed the effect of oral LA supplementation on flow-mediated vasodilation in 58 patients diagnosed with metabolic syndrome, a condition characterized by abnormal glucose and lipid (fat) metabolism.56 Oral supplementation with 300 mg/day of LA for 4 weeks improved flow-mediated vasodilation by 44% compared with placebo. Patients with diabetes are also at high risk for microvascular disease, which may contribute to diabetic neuropathy.45 In an uncontrolled study, oral supplementation with 1200 mg/day of racemic LA for 6 weeks improved a measure of capillary perfusion in the fingers of eight patients with diabetes who had peripheral neuropathy.57 While these results are encouraging, long-term randomized controlled trials are needed to determine whether LA supplementation can reduce the risk of vascular complications in individuals with diabetes.

Diabetic Neuropathy

More than 20% of patients with diabetes develop peripheral neuropathy, a type of nerve damage that may result in pain, loss of sensation, and weakness, particularly in the lower extremities.45 Peripheral neuropathy is also a leading cause of lower-limb amputation in patients with diabetes.58 Chronic hyperglycemia has been linked to peripheral nerve damage; several mechanisms have been proposed to explain the glucose-induced nerve damage, such as intracellular accumulation of sorbitol, glycation reactions, and oxidative and nitrosative stress.59 The results of several large randomized controlled trials indicate that maintaining blood glucose at near normal levels is the most important step in decreasing the risk of diabetic neuropathy.60,61 However, such intensive blood glucose control may not be achievable in all patients with diabetes.

Intravenous and oral LA are approved for the treatment of diabetic neuropathy in Germany.4 A meta-analysis that combined the results of four randomized controlled trials, including 1258 patients with diabetes, found that treatment with 600 mg/day of intravenous racemic LA for 3 weeks significantly reduced the symptoms of diabetic neuropathy to a clinically meaningful degree.62

The efficacy of oral LA in the treatment of diabetic neuropathy is less clear. A short-term study of 24 patients with type 2 DM found that the symptoms of peripheral neuropathy were improved in those who took 1800 mg/day of oral racemic LA for 3 weeks compared with those who took a placebo.63 More recently, a randomized, double-blind, placebo-controlled trial in 181 patients with diabetic neuropathy found that oral supplementation with 600 mg/day, 1200 mg/day, or 1800 mg/day of racemic LA for 5 weeks significantly improved neuropathic symptoms.64 In this study, the 600 mg/day dose was as effective as the higher doses. A much larger clinical trial randomly assigned more than 500 patients with type 2 DM and symptomatic peripheral neuropathy to one of the following treatments: (1) 600 mg/day of intravenous racemic LA for 3 weeks, followed by 1800 mg/day of oral racemic LA for 6 months; (2) 600 mg/day of intravenous racemic LA for 3 weeks followed by oral placebo for 6 months; or (3) intravenous placebo for 3 weeks followed by oral placebo for 6 months.65 Although symptom scores did not differ significantly from baseline in any of the groups, physician assessments of sensory and motor deficits improved significantly after 3 weeks of intravenous LA therapy. Motor and sensory deficits were also somewhat improved at the end of 6 months of oral LA therapy, but the trend did not reach statistical significance. In another trial of oral LA therapy, 299 patients with diabetic peripheral neuropathy were randomly assigned to treatment with 1200 mg/day of racemic LA, 600 mg/day of racemic LA, or a placebo.66 However, after 2 years of treatment, only 65 of the original participants were included in the final analysis. In that subgroup, those who took either 1200 mg/day or 600 mg/day of LA showed significant improvement in electrophysiological tests of nerve conduction compared with those who took the placebo. In the longest clinical trial of oral LA therapy, 421 patients with diabetes and distal symmetric sensorimotor polyneuropathy took either 600 mg/day of racemic LA or a placebo for 4 years.67 No difference between the two groups was seen for the primary endpoint, a composite score that assessed neuropathic impairment of the lower limbs and nerve conduction; however, some measures of neuropathic impairment improved with LA supplementation.

Another neuropathic complication of diabetes is cardiovascular autonomic neuropathy (CAN), which occurs in as many as 25% of patients with diabetes.45 CAN is characterized by reduced heart-rate variability (HRV; variability in the time interval between heartbeats) and is associated with increased risk of mortality in patients with diabetes. In a randomized controlled trial of 72 patients with type 2 DM and reduced HRV, oral supplementation with 800 mg/day of racemic LA for 4 months resulted in significant improvement in two out of four measures of HRV compared with placebo.68

Overall, the available research suggests that treatment with 600 mg/day of intravenous LA for 3 weeks significantly reduces the symptoms of diabetic peripheral neuropathy. Although the benefit of long-term oral LA supplementation is less clear, there is some evidence to suggest that oral LA may be beneficial in the treatment of diabetic peripheral neuropathy (600–1800 mg/day) and cardiovascular autonomic neuropathy (800 mg/day).

Multiple Sclerosis

Feeding high doses of LA to mice with experimental autoimmune encephalomyelitis, a model of multiple sclerosis (MS), has been found to slow disease progression.69,70 LA treatment through subcutaneous injection also reduced clinical signs of the disease in a rat model of MS.71 LA treatment has been shown to inhibit the migration of leukocytes (inflammatory T cells, monocytes, and macrophages) into the brain and spinal cord, possibly by decreasing endothelial expression of cell adhesion molecules, inhibiting enzymes called matrix metalloproteinases (MMPs), and reducing the permeability of the blood–brain barrier.69,71–73 More recently, LA has been found to reduce the production of proinflammatory cytokines74 and stimulate the production of cyclic adenosine monophosphate (AMP) and cell signaling in certain immune cells,74,75 which may also modulate the effects of LA in MS.

Although the results of animal studies are promising, human research is needed to determine whether oral LA supplementation might be efficacious in MS. A small pilot study designed to evaluate the safety of LA in 30 people with relapsing or progressive MS found that treatment with 1200–2400 mg/day of oral LA for 2 weeks was generally well tolerated (see the Safety section below), and that higher peak serum levels of LA were associated with greater decreases in serum MMP-9 levels.76 A pharmacokinetic study showed that an oral dose of 1200 mg of racemic LA can result in similar serum levels in MS patients as those found to be therapeutic in mice.77 However, large-scale, long-term clinical trials are needed to assess the safety and efficacy of LA in the treatment of MS.78

Cognitive Decline and Dementia

LA, alone or in combination with other antioxidants or L-carnitine, has been found to improve measures of memory in aged animals or in animal models of age-associated cognitive decline, including rats,79,80 mice,8184 and dogs.85 Memory assessments were done at the end of the LA treatment period, and it is not known whether LA treatment might have lasting memory effects in these animal models. It is also not clear whether oral LA supplementation can slow cognitive decline related to aging or pathological conditions in humans. An uncontrolled, open-label trial in nine patients with probable Alzheimer disease and related dementias, who were taking acetylcholinesterase inhibitors, reported that oral supplementation with 600 mg/day of racemic LA appeared to stabilize cognitive function over a 1-year period.86 This study was subsequently extended to include 43 patients with probable Alzheimer disease, who were followed up to 4 years. Patients with mild dementia or moderate-early dementia who took 600 mg/day of racemic LA, in addition to acetylcholinesterase inhibitors, experienced slower cognitive decline compared with the typical cognitive decline of patients with Alzheimer disease as reported in the literature.87 However, the significance of these findings is difficult to assess without a control group for comparison. A randomized controlled trial found that oral supplementation with 1200 mg/day of racemic LA for 10 weeks was of no benefit in treating HIV-associated cognitive impairment.88 Although studies in animals suggest that LA may be helpful in slowing age-related cognitive decline, randomized controlled trials are needed to determine whether LA supplementation is effective in preventing or slowing cognitive decline associated with age or neurodegenerative diseases.

Sources

Endogenous Biosynthesis

R-LA is synthesized endogenously by humans and bound to proteins (see the Bioavailability and Metabolism section above).

Food Sources

R-LA occurs naturally in foods covalently bound to lysine in proteins (lipoyllysine). Although LA is found in a wide variety of foods from plant and animal sources, quantitative information on the LA or lipoyllysine content of food is limited and published databases are lacking. Animal tissues that are rich in lipoyllysine (approx. 1–3 µg/g dry weight) include kidney, heart, and liver, while vegetables that are rich in lipoyllysine include spinach and broccoli.89 Somewhat lower amounts of lipoyllysine (approx. 0.5 µg/g dry weight) have been measured in tomatoes, peas, and Brussels sprouts.

Supplements

Unlike LA in foods, LA in supplements is free, meaning it is not bound to protein. Moreover, the amounts of LA available in dietary supplements (200–600 mg) are likely as much as 1000 times greater than the amounts that could be obtained in the diet. In Germany, LA is approved for the treatment of diabetic neuropathies and is available by prescription.44 LA is available as a dietary supplement without a prescription in the United States.90 Most LA supplements contain a racemic (50/50) mixture of R-LA and S-LA (d,l-LA). Supplements that claim to contain only R-LA are usually more expensive, and information regarding their purity is not currently available.91 Since taking LA with a meal decreases its bioavailability, it is generally recommended that LA be taken on an empty stomach (1 hour before or 2 hours after eating).

Racemic versus R-LA Supplements

R-LA is the isomer that is synthesized by plants and animals and functions as a cofactor for mitochondrial enzymes in its protein-bound form (see the Biological Activities section above). Direct comparisons of the bioavailability of oral racemic LA and R-LA supplements have not been published. After oral dosing with racemic LA, peak plasma concentrations of R-LA were found to be 40%–50% higher than S-LA, suggesting R-LA is better absorbed than S-LA, but both isomers are rapidly metabolized and eliminated.11,13,16 In rats, R-LA was more effective than S-LA in enhancing insulin-stimulated glucose transport and metabolism in skeletal muscle,49 and R-LA was more effective than racemic LA and S-LA in preventing cataracts.92 However, virtually all of the published studies of LA supplementation in humans have used racemic LA. At present, it is not clear whether R-LA supplements are more effective than racemic LA supplements in humans.

Safety

Adverse Effects

In general, LA supplementation at moderate doses has been found to have few serious side effects. When used to treat diabetic peripheral neuropathy, intravenous administration of racemic LA at doses of 600 mg/day for 3 weeks62 and oral racemic LA at doses as high as 1800 mg/day for 6 months66 and 1200 mg/day for 2 years65 did not result in serious adverse effects. Two mild anaphylactoid reactions and one severe anaphylactic reaction, including laryngospasm, were reported after intravenous LA administration.45 The most frequently reported side effects of oral LA supplementation are allergic reactions affecting the skin, including rashes, hives, and itching. Abdominal pain, nausea, vomiting, diarrhea, and vertigo have also been reported, and one trial found that the incidence of nausea, vomiting, and vertigo was dose dependent.64 Further, malodorous urine has been noted by people taking 1200 mg/day of LA orally.76

Pregnancy and Lactation

The safety of LA supplements in pregnant and lactating women has not been established.93

Drug Interactions

Because there is some evidence that LA supplementation improves insulin-mediated glucose utilization,48 it is possible that LA supplementation could increase the risk of hypoglycemia in patients with diabetes who are using insulin or oral antidiabetic agents. Consequently, blood glucose levels should be monitored closely when LA supplementation is added to diabetes treatment regimens. Co-administration of a single oral dose of racemic LA (600 mg) and the oral antidiabetic agents, glyburide or acarbose, did not result in any significant drug interactions in one study in 24 healthy volunteers.94

Nutrient Interactions Biotin

References

The chemical structure of biotin is similar to that of LA, and there is some evidence that high concentrations of LA can compete with biotin for transport across cell membranes.95,96 The administration of high doses of LA by injection to rats decreased the activity of two biotin-dependent enzymes by approximately 30%–35%,97 but it is not known whether oral or intravenous LA supplementation substantially increases the requirement for biotin in humans.98

Summary

α-Lipoic acid (LA), also known as thioctic acid, is a naturally occurring compound that is synthesized in small amounts by humans.

Endogenously synthesized LA is bound to protein and functions as a cofactor for several important mitochondrial enzymes.

Supplementation with high doses of LA transiently increases plasma and cellular levels of free LA.

Although LA is a potent antioxidant in the test tube, LA supplementation may affect health by stimulating glutathione synthesis, enhancing insulin signaling, and modulating the activity of other cell-signaling molecules and transcription factors.

Overall, the available research indicates that treatment with 600 mg/day of intravenous racemic LA for 3 weeks significantly reduces the symptoms of diabetic peripheral neuropathy.

Compared with intravenous administration, the effect of long-term, oral LA supplementation for diabetic peripheral neuropathy is less clear, yet some studies show that oral supplementation with at least 600 mg/day of racemic LA may be beneficial.

It is not yet known whether LA supplementation is beneficial in the treatment of multiple sclerosis or neurodegenerative diseases like Alzheimer disease.

For those who choose to take LA supplements, the Linus Pauling Institute recommends a daily dose of 200–400 mg/day of racemic LA for generally healthy people.

References

1. Reed LJ. A trail of research from lipoic acid to alphaketo acid dehydrogenase complexes. J Biol Chem 2001;276(42):38329–38336

2. Carreau JP. Biosynthesis of lipoic acid via unsaturated fatty acids. Methods Enzymol 1979;62:152–158

3. Smith AR, Shenvi SV, Widlansky M, Suh JH, Hagen TM. Lipoic acid as a potential therapy for chronic diseases associated with oxidative stress. Curr Med Chem 2004;11(9):1135–1146

4. Kramer K, Packer L. R-alpha-lipoic acid. In: Kramer K, Hoppe P, Packer L, eds. Nutraceuticals in Health and Disease Prevention. New York: Marcel Dekker, Inc; 2001:129–164

5. Cicchillo RM, Iwig DF, Jones AD, et al. Lipoyl synthase requires two equivalents of S-adenosyl-L-methionine to synthesize one equivalent of lipoic acid. Biochemistry 2004;43(21):6378–6386

6. Zhao X, Miller JR, Jiang Y, Marletta MA, Cronan JE. Assembly of the covalent linkage between lipoic acid and its cognate enzymes. Chem Biol 2003; 10(12): 1293–1302

7. Cicchillo RM, Booker SJ. Mechanistic investigations of lipoic acid biosynthesis in Escherichia coli: both sulfur atoms in lipoic acid are contributed by the same lipoyl synthase polypeptide. J Am Chem Soc 2005; 127(9):2860–2861

8. Miller JR, Busby RW, Jordan SW, et al. Escherichia coli LipA is a lipoyl synthase: in vitro biosynthesis of lipoylated pyruvate dehydrogenase complex from octanoyl-acyl carrier protein. Biochemistry 2000; 39(49):15166–15178

9. Fujiwara K, Suzuki M, Okumachi Y, et al. Molecular cloning, structural characterization and chromosomal localization of human lipoyltransferase gene. Eur J Biochem 1999;260(3):761–767

10. Fujiwara K, Takeuchi S, Okamura-Ikeda K, Motokawa Y. Purification, characterization, and cDNA cloning of lipoate-activating enzyme from bovine liver. J Biol Chem 2001;276(31):28819–28823

11. Hermann R, Niebch G, Borbe H, et al. Enantioselective pharmacokinetics and bioavailability of different racemic alpha-lipoic acid formulations in healthy volunteers. Eur J Pharm Sci 1996;4(3):167–174

12. Teichert J, Hermann R, Ruus P, Preiss R. Plasma kinetics, metabolism, and urinary excretion of alpha-lipoic acid following oral administration in healthy volunteers. J Clin Pharmacol 2003;43(11):1257–1267

13. Gleiter CH, Schug BS, Hermann R, Elze M, Blume HH, Gundert-Remy U. Influence of food intake on the bioavailability of thioctic acid enantiomers. Eur J Clin Pharmacol 1996;50(6):513–514

14. Carlson DA, Smith AR, Fischer SJ, Young KL, Packer L. The plasma pharmacokinetics of R-(+)-lipoic acid administered as sodium R-(+)-lipoate to healthy human subjects. Altern Med Rev 2007;12(4):343–351

15. Evans JL, Heymann CJ, Goldfine ID, Gavin LA. Pharmacokinetics, tolerability, and fructosamine-lowering effect of a novel, controlled-release formulation of alpha-lipoic acid. Endocr Pract 2002;8(1):29–35

16. Breithaupt-Grögler K, Niebch G, Schneider E, et al. Dose-proportionality of oral thioctic acid—coincidence of assessments via pooled plasma and individual data. Eur J Pharm Sci 1999;8(1):57–65

17. Bustamante J, Lodge JK, Marcocci L, Tritschler HJ, Packer L, Rihn BH. Alpha-lipoic acid in liver metabolism and disease. Free Radic Biol Med 1998; 24(6): 1023–1039

18. Harris RA, Joshi M, Jeoung NH, Obayashi M. Overview of the molecular and biochemical basis of branchedchain amino acid catabolism. J Nutr 2005; 135(6, Suppl):1527S–1530S

19. Packer L. alpha-Lipoic acid: a metabolic antioxidant which regulates NF-kappa B signal transduction and protects against oxidative injury. Drug Metab Rev 1998;30(2):245–275

20. Jones W, Li X, Qu ZC, Perriott L, Whitesell RR, May JM. Uptake, recycling, and antioxidant actions of alpha-lipoic acid in endothelial cells. Free Radic Biol Med 2002;33(1):83–93

21. May JM, Qu ZC, Mendiratta S. Protection and recycling of alphatocopherol in human erythrocytes by intracellular ascorbic acid. Arch Biochem Biophys 1998;349(2):281–289

22. Kozlov AV, Gille L, Staniek K, Nohl H. Dihydrolipoic acid maintains ubiquinone in the antioxidant active form by two-electron reduction of ubiquinone and one-electron reduction of ubisemiquinone. Arch Biochem Biophys 1999;363(1):148–154

23. Upston JM, Terentis AC, Stocker R. Tocopherol-mediated peroxidation of lipoproteins: implications for vitamin E as a potential antiatherogenic supplement. FASEB J 1999;13(9):977–994

24. Valko M, Morris H, Cronin MT. Metals, toxicity and oxidative stress. Curr Med Chem 2005;12(10):1161–1208

25. Doraiswamy PM, Finefrock AE. Metals in our minds: therapeutic implications for neurodegenerative disorders. Lancet Neurol 2004;3(7):431–434

26. Ou P, Tritschler HJ, Wolff SP. Thioctic (lipoic) acid: a therapeutic metal-chelating antioxidant? Biochem Pharmacol 1995;50(1):123–126

27. Suh JH, Zhu BZ, deSzoeke E, Frei B, Hagen TM. Dihydrolipoic acid lowers the redox activity of transition metal ions but does not remove them from the active site of enzymes. Redox Rep 2004;9(1):57–61

28. Yamamoto H, Watanabe T, Mizuno H, et al. The antioxidant effect of DL-alpha-lipoic acid on copper-induced acute hepatitis in Long-Evans Cinnamon (LEC) rats. Free Radic Res 2001;34(1):69–80

29. Suh JH, Moreau R, Heath SH, Hagen TM. Dietary supplementation with (R)-alpha-lipoic acid reverses the age-related accumulation of iron and depletion of antioxidants in the rat cerebral cortex. Redox Rep 2005;10(1):52–60

30. Hagen TM, Vinarsky V, Wehr CM, Ames BN. (R)-alpha-lipoic acid reverses the age-associated increase in susceptibility of hepatocytes to tert-butylhydroper-oxide both in vitro and in vivo. Antioxid Redox Signal 2000;2(3):473–483

31. Busse E, Zimmer G, Schopohl B, Kornhuber B. Influence of alpha-lipoic acid on intracellular glutathione in vitro and in vivo. Arzneimittelforschung 1992; 42(6):829–831

32. Monette JS, Gómez LA, Moreau RF, et al. (R)-α-Lipoic acid treatment restores ceramide balance in aging rat cardiac mitochondria. Pharmacol Res 2011;63(1):23–29

33. Suh JH, Shenvi SV, Dixon BM, et al. Decline in transcriptional activity of Nrf2 causes age-related loss of glutathione synthesis, which is reversible with lipoic acid. Proc Natl Acad Sci U S A 2004;101(10):3381–3386

34. Suh JH, Wang H, Liu RM, Liu J, Hagen TM. (R)-alpha-lipoic acid reverses the age-related loss in GSH redox status in postmitotic tissues: evidence for increased cysteine requirement for GSH synthesis. Arch Biochem Biophys 2004;423(1):126–135

35. Konrad D. Utilization of the insulin-signaling network in the metabolic actions of alpha-lipoic acid-reduction or oxidation? Antioxid Redox Signal 2005;7(7–8):1032–1039

36. Diesel B, Kulhanek-Heinze S, Höltje M, et al. Alpha-lipoic acid as a directly binding activator of the insulin receptor: protection from hepatocyte apoptosis. Biochemistry 2007;46(8):2146–2155

37. Yaworsky K, Somwar R, Ramlal T, Tritschler HJ, Klip A. Engagement of the insulin-sensitive pathway in the stimulation of glucose transport by alpha-lipoic acid in 3T3-L1 adipocytes. Diabetologia 2000;43(3):294–303

38. Estrada DE, Ewart HS, Tsakiridis T, et al. Stimulation of glucose uptake by the natural coenzyme alpha-lipoic acid/thioctic acid: participation of elements of the insulin signaling pathway. Diabetes 1996;45(12): 1798–1804

39. Zhang L, Xing GQ, Barker JL, et al. Alpha-lipoic acid protects rat cortical neurons against cell death induced by amyloid and hydrogen peroxide through the Akt signalling pathway. Neurosci Lett 2001;312(3): 125–128

40. Shay KP, Hagen TM. Age-associated impairment of Akt phosphorylation in primary rat hepatocytes is remediated by alpha-lipoic acid through PI3 kinase, PTEN, and PP2A. Biogerontology 2009;10(4):443–456

41. Zhang WJ, Wei H, Hagen T, Frei B. Alpha-lipoic acid attenuates LPS-induced inflammatory responses by activating the phosphoinositide 3-kinase/Akt signaling pathway. Proc Natl Acad Sci U S A 2007;104(10): 4077–4082

42. Smith AR, Hagen TM. Vascular endothelial dysfunction in aging: loss of Akt-dependent endothelial nitric oxide synthase phosphorylation and partial restoration by (R)-alpha-lipoic acid. Biochem Soc Trans 2003;31(Pt 6):1447–1449

43. Zhang WJ, Frei B. Alpha-lipoic acid inhibits TNF-alpha-induced NF-kappaB activation and adhesion molecule expression in human aortic endothelial cells. FASEB J 2001;15(13):2423–2432

44. Biewenga GP, Haenen GR, Bast A. The pharmacology of the antioxidant lipoic acid. Gen Pharmacol 1997; 29(3):315–331

45. Ziegler D. Thioctic acid for patients with symptomatic diabetic polyneuropathy: a critical review. Treat Endocrinol 2004;3(3):173–189

46. Jacob S, Henriksen EJ, Schiemann AL, et al. Enhancement of glucose disposal in patients with type 2 diabetes by alpha-lipoic acid. Arzneimittelforschung 1995;45(8):872–874

47. Jacob S, Henriksen EJ, Tritschler HJ, Augustin HJ, Dietze GJ. Improvement of insulin-stimulated glucose-disposal in type 2 diabetes after repeated parenteral administration of thioctic acid. Exp Clin Endocrinol Diabetes 1996;104(3):284–288

48. Jacob S, Rett K, Henriksen EJ, Häring HU. Thioctic acid—effects on insulin sensitivity and glucose-metabolism. Biofactors 1999;10(2–3):169–174

49. Streeper RS, Henriksen EJ, Jacob S, Hokama JY, Fogt DL, Tritschler HJ. Differential effects of lipoic acid stereoisomers on glucose metabolism in insulin-resistant skeletal muscle. Am J Physiol 1997;273(1 Pt 1): E185–E191

50. Schalkwijk CG, Stehouwer CD. Vascular complications in diabetes mellitus: the role of endothelial dysfunction. Clin Sci (Lond) 2005;109(2):143–159

51. Heitzer T, Finckh B, Albers S, Krohn K, Kohlschütter A, Meinertz T. Beneficial effects of alpha-lipoic acid and ascorbic acid on endothelium-dependent, nitric oxide-mediated vasodilation in diabetic patients: relation to parameters of oxidative stress. Free Radic Biol Med 2001;31(1):53–61

52. Heinisch BB, Francesconi M, Mittermayer F, et al. Alpha-lipoic acid improves vascular endothelial function in patients with type 2 diabetes: a placebo-controlled randomized trial. Eur J Clin Invest 2010; 40(2):148–154

53. Gokce N, Keaney JF Jr, Hunter LM, Watkins MT, Menzoian JO, Vita JA. Risk stratification for postoperative cardiovascular events via noninvasive assessment of endothelial function: a prospective study. Circulation 2002;105(13):1567–1572

54. Xiang G, Pu J, Yue L, Hou J, Sun H. α-lipoic acid can improve endothelial dysfunction in subjects with impaired fasting glucose. Metabolism 2011;60(4):480–485

55. Xiang GD, Sun HL, Zhao LS, Hou J, Yue L, Xu L. The antioxidant alpha-lipoic acid improves endothelial dysfunction induced by acute hyperglycaemia during OGTT in impaired glucose tolerance. Clin Endocrinol (Oxf) 2008;68(5):716–723

56. Sola S, Mir MQ, Cheema FA, et al. Irbesartan and lipoic acid improve endothelial function and reduce markers of inflammation in the metabolic syndrome: results of the Irbesartan and Lipoic Acid in Endothelial Dysfunction (ISLAND) study. Circulation 2005; 111(3):343–348

57. Haak E, Usadel KH, Kusterer K, et al. Effects of alpha-lipoic acid on microcirculation in patients with peripheral diabetic neuropathy. Exp Clin Endocrinol Diabetes 2000;108(3):168–174

58. Greene DA, Stevens MJ, Obrosova I, Feldman EL. Glucose-induced oxidative stress and programmed cell death in diabetic neuropathy. Eur J Pharmacol 1999;375(1–3):217–223

59. Obrosova IG. Diabetes and the peripheral nerve. Biochim Biophys Acta 2009;1792(10):931–940

60. The Diabetes Control and Complications Trial Research Group. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med 1993;329(14):977–986

61. UK Prospective Diabetes Study (UKPDS) Group. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet 1998;352(9131):837–853

62. Ziegler D, Nowak H, Kempler P, Vargha P, Low PA. Treatment of symptomatic diabetic polyneuropathy with the antioxidant alpha-lipoic acid: a meta-analysis. Diabet Med 2004;21(2):114–121

63. Ruhnau KJ, Meissner HP, Finn JR, et al. Effects of 3-week oral treatment with the antioxidant thioctic acid (alpha-lipoic acid) in symptomatic diabetic polyneuropathy. Diabet Med 1999;16(12):1040–1043

64. Ziegler D, Ametov A, Barinov A, et al. Oral treatment with alpha-lipoic acid improves symptomatic diabetic polyneuropathy: the SYDNEY 2 trial. Diabetes Care 2006;29(11):2365–2370

65. Ziegler D, Hanefeld M, Ruhnau KJ, et al. Treatment of symptomatic diabetic polyneuropathy with the antioxidant alpha-lipoic acid: a 7-month multicenter randomized controlled trial (ALADIN III Study). ALADIN III Study Group. Alpha-Lipoic Acid in Diabetic Neuropathy. Diabetes Care 1999;22(8):1296–1301

66. Reljanovic M, Reichel G, Rett K, et al. Treatment of diabetic polyneuropathy with the antioxidant thioctic acid (alpha-lipoic acid): a two year multicenter randomized double-blind placebo-controlled trial (ALADIN II). Alpha Lipoic Acid in Diabetic Neuropathy. Free Radic Res 1999;31(3):171–179

67. Ziegler D, Low PA, Litchy WJ, et al. Efficacy and safety of antioxidant treatment with α-lipoic acid over 4 years in diabetic polyneuropathy: the NATHAN 1 trial. Diabetes Care 2011;34(9):2054–2060

68. Ziegler D, Schatz H, Conrad F, Gries FA, Ulrich H, Reichel G. Effects of treatment with the antioxidant alpha-lipoic acid on cardiac autonomic neuropathy in NIDDM patients. A 4-month randomized controlled multicenter trial (DEKAN Study). Deutsche Kardiale Autonome Neuropathie. Diabetes Care 1997;20(3): 369–373

69. Marracci GH, Jones RE, McKeon GP, Bourdette DN. Alpha lipoic acid inhibits T cell migration into the spinal cord and suppresses and treats experimental autoimmune encephalomyelitis. J Neuroimmunol 2002; 131(1–2):104–114

70. Morini M, Roccatagliata L, Dell'Eva R, et al. Alpha-lipoic acid is effective in prevention and treatment of experimental autoimmune encephalomyelitis. J Neuroimmunol 2004;148(1–2):146–153

71. Schreibelt G, Musters RJ, Reijerkerk A, et al. Lipoic acid affects cellular migration into the central nervous system and stabilizes blood-brain barrier integrity. J Immunol 2006;177(4):2630–2637

72. Chaudhary P, Marracci GH, Bourdette DN. Lipoic acid inhibits expression of ICAM-1 and VCAM-1 by CNS endothelial cells and T cell migration into the spinal cord in experimental autoimmune encephalomyelitis. J Neuroimmunol 2006;175(1–2):87–96

73. Marracci GH, McKeon GP, Marquardt WE, Winter RW, Riscoe MK, Bourdette DN. Alpha lipoic acid inhibits human T-cell migration: implications for multiple sclerosis. J Neurosci Res 2004;78(3):362–370

74. Salinthone S, Schillace RV, Tsang C, Regan JW, Bourdette DN, Carr DW. Lipoic acid stimulates cAMP production via G protein-coupled receptor-dependent and -independent mechanisms. J Nutr Biochem 2011;22(7):681–690

75. Schillace RV, Pisenti N, Pattamanuch N, et al. Lipoic acid stimulates cAMP production in T lymphocytes and NK cells. Biochem Biophys Res Commun 2007; 354(1):259–264

76. Yadav V, Marracci G, Lovera J, et al. Lipoic acid in multiple sclerosis: a pilot study. Mult Scler 2005; 11(2):159–165

77. Yadav V, Marracci GH, Munar MY, et al. Pharmacokinetic study of lipoic acid in multiple sclerosis: comparing mice and human pharmacokinetic parameters. Mult Scler 2010;16(4):387–397

78. National Multiple Sclerosis Society. Progress in Research: Research Highlights Winter/Spring 2005. Available at: http://nationalmssociety.org/Highlights-Antioxidants.asp. Accessed Jan 9, 2006

79. Liu J, Head E, Gharib AM, et al. Memory loss in old rats is associated with brain mitochondrial decay and RNA/DNA oxidation: partial reversal by feeding acetyl-L-carnitine and/or R-alpha -lipoic acid. Proc Natl Acad Sci U S A 2002;99(4):2356–2361

80. Hagen TM, Liu J, Lykkesfeldt J, et al. Feeding acetyl-Lcarnitine and lipoic acid to old rats significantly improves metabolic function while decreasing oxidative stress. Proc Natl Acad Sci U S A 2002;99(4):1870–1875

81. Farr SA, Poon HF, Dogrukol-Ak D, et al. The antioxidants alpha-lipoic acid and N-acetylcysteine reverse memory impairment and brain oxidative stress in aged SAMP8 mice. J Neurochem 2003;84(5):1173–1183

82. Quinn JF, Bussiere JR, Hammond RS, et al. Chronic dietary alpha-lipoic acid reduces deficits in hippocampal memory of aged Tg2576 mice. Neurobiol Aging 2007;28(2):213–225

83. Shenk JC, Liu J, Fischbach K, et al. The effect of acetyl-L-carnitine and R-alpha-lipoic acid treatment in ApoE4 mouse as a model of human Alzheimer's disease. J Neurol Sci 2009;283(1–2):199–206

84. Stoll S, Hartmann H, Cohen SA, Müller WE. The potent free radical scavenger alpha-lipoic acid improves memory in aged mice: putative relationship to NMDA receptor deficits. Pharmacol Biochem Behav 1993; 46(4):799–805

85. Milgram NW, Head E, Zicker SC, et al. Learning ability in aged beagle dogs is preserved by behavioral enrichment and dietary fortification: a two-year longitudinal study. Neurobiol Aging 2005;26(1):77–90

86. Hager K, Marahrens A, Kenklies M, Riederer P, Münch G. Alpha-lipoic acid as a new treatment option for Alzheimer [corrected] type dementia. Arch Gerontol Geriatr 2001;32(3):275–282

87. Hager K, Kenklies M, McAfoose J, Engel J, Münch G. Alpha-lipoic acid as a new treatment option for Alzheimer's disease—a 48 months follow-up analysis. J Neural Transm Suppl 2007; (72):189–193

88. Dana Consortium on the Therapy of HIV Dementia and Related Cognitive Disorders. A randomized, double-blind, placebo-controlled trial of deprenyl and thioctic acid in human immunodeficiency virus-associated cognitive impairment. Neurology 1998; 50(3): 645–651

89. Lodge JK, Youn HD, Handelman GJ, et al. Natural sources of lipoic acid: determination of lipoyllysine released from protease-digested tissues by high performance liquid chromatography incorporating electrochemical detection. J Appl Nutr 1997;49(1 & 2): 3–11

90. Hendler SS, Rorvik DR, eds. PDR for Nutritional Supplements. Montbale, NJ: Medical Economics Company, Inc; 2001

91. Alpha-Lipoic Acid Supplements. ConsumerLab.com. October 2, 2009. Available at: https://www.consumerlab.com/reviews/Alpha-Lipoic_Acid_Supplements/alphalipoic/. Accessed May 10, 2012

92. Maitra I, Serbinova E, Tritschler HJ, Packer L. Stereo-specific effects of R-lipoic acid on buthionine sulfoximine-induced cataract formation in newborn rats. Biochem Biophys Res Commun 1996;221(2):422–429

93. Hendler SS, Rorvik DR. Alpha-Lipoic Acid. In: Hendler SS, Rorvik DR, eds. PDR for Nutritional Supplements. 2nd ed. Montvale, NJ: Physicians’ Desk Reference Inc; 2008:25–29

94. Gleiter CH, Schreeb KH, Freudenthaler S, et al. Lack of interaction between thioctic acid, glibenclamide and acarbose. Br J Clin Pharmacol 1999;48(6):819–825

95. Prasad PD, Wang H, Huang W, et al. Molecular and functional characterization of the intestinal Na+-dependent multivitamin transporter. Arch Biochem Biophys 1999;366(1):95–106

96. Balamurugan K, Vaziri ND, Said HM. Biotin uptake by human proximal tubular epithelial cells: cellular and molecular aspects. Am J Physiol Renal Physiol 2005;288(4):F823–F831

97. Zempleni J, Trusty TA, Mock DM. Lipoic acid reduces the activities of biotin-dependent carboxylases in rat liver. J Nutr 1997;127(9):1776–1781

98. Zempleni J, Mock DM. Biotin biochemistry and human requirements. J Nutr Biochem 1999;10(3):128–138